J Bacteriol Virol.  2019 Mar;49(1):12-26. 10.4167/jbv.2019.49.1.12.

Effector Pathways of Toll-like Receptor-inducible Innate Immune Responses in Macrophages

Affiliations
  • 1Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea 35015. hayoungj@cnu.ac.kr
  • 2Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea 35015.

Abstract

Toll-like receptors (TLR) are well-characterized pattern recognition receptors that can recognize and respond to diverse pathogen-associated or danger-associated molecular patterns during infection. TLR signaling in macrophages triggers in the intracellular signaling pathways through the recruitment of various adaptor and signaling proteins, and results in the activation of effector mechanisms and pathways that are important for host defense to intracellular bacteria. Effector mechanisms include inflammatory responses, cytokine generation, production of reactive oxygen species, and antimicrobial proteins. Accumulating studies showed that autophagy is a key pathway in the maintenance of homeostasis and housekeeping functions during infection and inflammation. In this review, we summarize the major effector pathways and mechanisms in the activation of TLR-inducible innate immune responses in macrophages. In addition, we focus the emerging evidence of crosstalk between autophagy and TLR-mediated signaling in terms of effector function of innate immune responses. A better understanding of effector functions by the activation of TLR-mediated signaling cascades contributes to the development of new therapeutics and vaccines against various intracellular pathogenic infections.

Keyword

TLR; Cytokine; Antimicrobial Protein; Effectors; Autophagy; Innate Immunity

MeSH Terms

Autophagy
Bacteria
Homeostasis
Housekeeping
Immunity, Innate*
Inflammation
Macrophages*
Reactive Oxygen Species
Receptors, Pattern Recognition
Toll-Like Receptors
Vaccines
Reactive Oxygen Species
Receptors, Pattern Recognition
Toll-Like Receptors
Vaccines

Figure

  • Figure 1 An overview of TLR-induced intracellular signaling pathways in innate immune cells. Upon TLR signaling, the intracellular signaling cascades involve the actions of numerous adaptors MyD88, TRIF, MAL/TIRARAP, or TRAM, and multiple signaling molecules. In particular, MyD88 is recruited by all TLR molecules, whereas TRIF is utilized by TLR3 and 4. The recruitment of MyD88 activates IRAK kinase family members, which associates with E3 ubiquitin ligase TRAF6 to activate TAK1 protein kinase complex. This leads to the activation of both NF-κB and mitogen-activated protein kinase (MAPK) pathways (ERK1/2, p38, and JNK) for proinflammatory cytokine generation. The other adaptor TRIF interacts with both TRAF6 and TRAF3, to facilitate the activation of TAK1 complex and TBK1 pathways, leading to the activation of type I interferon responses. In the sophisticated process, uncoordinated 93 homolog B1 (UNC93B 1) and leucine-rich repeat containing protein (LRRC) 59, are required for the endosomal TLR trafficking from ER to endosomal structures.

  • Figure 2 Multiple functions of antimicrobial proteins in innate immune responses. TLR-induced intracellular signaling pathways culminate into the activation of innate effector pathways including the generation of antimicrobial proteins. Two important antimicrobial peptides, cathelicidins and defensins, function in various aspects of biological responses, i.e., the regulation of inflammation, phagocytosis, autophagy, as well as antimicrobial effects, in innate immune cells.

  • Figure 3 A schematic overview of macroautophagy and xenophagy in mycobacterial infection. Macroautophagy process include three steps: initiation (formation of phagophore), elongation into double-membraned autophagosomal structure, and its maturation by lysosomal fusion. Xenophagy process is well-characterized in mycobacterial infection. During Mtb infection, xenophagy is activated by cytoplasmic release of Mtb, which can be ubiquitinated by E3 ligases Parkin and Smurf1. Then ubiquitinated Mtb phagosomes are recognized by autophagic receptors p62 and NDP52, which contain domains for interaction with ubiquitinated cargos and LC3-containing autophagic machinery. It is also well-known that, several innate immune signals including TLR, vitamin D receptor signaling, IFN-γh distinct mechanisms involving IRGM (for IFN-γ), cathelicidins (for vitamin D receptor signaling), and TBK1 (for TLR) in monocytes/macrophages.

  • Figure 4 Figure 4. TLR-mediated regulation of antibacterial autophagy. TLR signaling activation in innate immune cells results in the activation of antibacterial autophagy. TLR7 signaling activates autophagy through MyD88; TLR4 stimulation induces autophagy through a TRIF-dependent pathway. TLR2/1 stimulation activates AMPK-dependent functional vitamin D signaling activation, leading to the induction of autophagy activation and antimicrobial responses. Either TLR2 or TLR4 stimulation activates autophagy through a serine protease inhibitor PAI-2 via stabilization of Beclin 1. In addition, TLR3, 7, 9, and UNC93B1, are required for the activation of antibacterial autophagy. TLR8 stimulation activates the expression of genes involved in vitamin D signaling and cathelicidin-dependent autophagy in human macrophages.


Reference

1. Akira S, Takeda K, Kaisho T. Toll-like receptors: critical proteins linking innate and acquired immunity. Nat Immunol. 2001; 2:675.
Article
2. Hu X, Chakravarty SD, Ivashkiv LB. Regulation of interferon and Toll-like receptor signaling during macrophage activation by opposing feedforward and feedback inhibition mechanisms. Immunol Rev. 2008; 226:41–56.
Article
3. Wynn TA, Barron L. Macrophages: master regulators of inflammation and fibrosis. Semin Liver Dis. 2010; 30:245–257.
Article
4. Cui J, Chen Y, Wang HY, Wang RF. Mechanisms and pathways of innate immune activation and regulation in health and cancer. Hum Vaccin Immunother. 2014; 10:3270–3285.
Article
5. Kawai T, Akira S. TLR signaling. Cell Death Differ. 2006; 13:816–825.
Article
6. Gulati A, Kaur D, Krishna Prasad GVR, Mukhopadhaya A. PRR function of innate immune receptors in recognition of bacteria or bacterial ligands. Adv Exp Med Biol. 2018; 1112:255–280.
Article
7. Vajjhala PR, Ve T, Bentham A, Stacey KJ, Kobe B. The molecular mechanisms of signaling by cooperative assembly formation in innate immunity pathways. Mol Immunol. 2017; 86:23–37.
Article
8. Kawasaki T, Kawai T. Toll-like receptor signaling pathways. Front Immunol. 2014; 5:461.
Article
9. Nanson JD, Kobe B, Ve T. Death, TIR, and RHIM: Self-assembling domains involved in innate immunity and cell-death signaling. J Leukoc Biol. 2019; 105:363–375.
Article
10. Campoy E, Colombo MI. Autophagy in intracellular bacterial infection. Biochim Biophys Acta. 2009; 1793:1465–1477.
Article
11. Borel S, Espert L, Biard-Piechaczyk M. Macroautophagy regulation during HIV-1 infection of CD4+ T cells and macrophages. Front Immunol. 2012; 3:97.
Article
12. Deretic V. Autophagy, an immunologic magic bullet: Mycobacterium tuberculosis phagosome maturation block and how to bypass it. Future Microbiol. 2008; 3:517–524.
Article
13. Bradfute SB, Castillo EF, Arko-Mensah J, Chauhan S, Jiang S, Mandell M, et al. Autophagy as an immune effector against tuberculosis. Curr Opin Microbiol. 2013; 16:355–365.
Article
14. Basu J, Shin DM, Jo EK. Mycobacterial signaling through toll-like receptors. Front Cell Infect Microbiol. 2012; 2:145.
Article
15. Sanjuan MA, Dillon CP, Tait SW, Moshiach S, Dorsey F, Connell S, et al. Toll-like receptor signalling in macrophages links the autophagy pathway to phagocytosis. Nature. 2007; 450:1253–1257.
Article
16. Deretic V, Levine B. Autophagy balances inflammation in innate immunity. Autophagy. 2018; 14:243–251.
Article
17. Deretic V, Saitoh T, Akira S. Autophagy in infection, inflammation and immunity. Nat Rev Immunol. 2013; 13:722–737.
Article
18. Fésüs L, Demény MÁ, Petrovski G. Autophagy shapes inflammation. Antioxid Redox Signal. 2011; 14:2233–2243.
Article
19. Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol. 2010; 11:373–384.
Article
20. Lee BL, Moon JE, Shu JH, Yuan L, Newman ZR, Schekman R, et al. UNC93B1 mediates differential trafficking of endosomal TLRs. Elife. 2013; 2:e00291.
Article
21. Tatematsu M, Funami K, Ishii N, Seya T, Obuse C, Matsumoto M. LRRC59 regulates trafficking of nucleic acid-sensing TLRs from the endoplasmic reticulum via association with UNC93B1. J Immunol. 2015; 195:4933–4942.
Article
22. Chen ZJ. Ubiquitination in signaling to and activation of IKK. Immunol Rev. 2012; 246:95–106.
Article
23. Ajibade AA, Wang HY, Wang RF. Cell type-specific function of TAK1 in innate immune signaling. Trends Immunol. 2013; 34:307–316.
Article
24. Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006; 124:783–801.
Article
25. Tseng PH, Matsuzawa A, Zhang W, Mino T, Vignali DA, Karin M. Different modes of ubiquitination of the adaptor TRAF3 selectively activate the expression of type I interferons and proinflammatory cytokines. Nat Immunol. 2010; 11:70–75.
Article
26. Zinngrebe J, Montinaro A, Peltzer N, Walczak H. Ubiquitin in the immune system. EMBO Rep. 2014; 15:28–45.
Article
27. Jiang X, Chen ZJ. The role of ubiquitylation in immune defence and pathogen evasion. Nat Rev Immunol. 2011; 12:35–48.
Article
28. Murphy M, Xiong Y, Pattabiraman G, Qiu F, Medvedev AE. Pellino-1 positively regulates toll-like receptor (TLR) 2 and TLR4 signaling and is suppressed upon induction of endotoxin tolerance. J Biol Chem. 2015; 290:19218–19232.
Article
29. Medvedev AE, Murphy M, Zhou H, Li X. E3 ubiquitin ligases Pellinos as regulators of pattern recognition receptor signaling and immune responses. Immunol Rev. 2015; 266:109–122.
Article
30. Wu C, Su Z, Lin M, Ou J, Zhao W, Cui J, et al. NLRP11 attenuates toll-like receptor signalling by targeting TRAF6 for degradation via the ubiquitin ligase RNF19A. Nat Commun. 2017; 8:1977.
Article
31. Lee EY, Lee MW, Wong GCL. Modulation of toll-like receptor signaling by antimicrobial peptides. Semin Cell Dev Biol. 2018; 88:173–184.
Article
32. Zhang LJ, Gallo RL. Antimicrobial peptides. Curr Biol. 2016; 26:R14–R19.
Article
33. Pasupuleti M, Schmidtchen A, Malmsten M. Antimicrobial peptides: key components of the innate immune system. Crit Rev Biotechnol. 2012; 32:143–171.
Article
34. Sung DK, Chang YS, Sung SI, Yoo HS, Ahn SY, Park WS. Antibacterial effect of mesenchymal stem cells against Escherichia coli is mediated by secretion of beta-defensin-2 via toll-like receptor 4 signalling. Cell Microbiol. 2016; 18:424–436.
Article
35. Wu YY, Hsu CM, Chen PH, Fung CP, Chen LW. Toll-like receptor stimulation induces nondefensin protein expression and reverses antibiotic-induced gut defense impairment. Infect Immun. 2014; 82:1994–2005.
Article
36. Pashenkov MV, Murugina NE, Budikhina AS, Pinegin BV. Synergistic interactions between NOD receptors and TLRs: Mechanisms and clinical implications. J Leukoc Biol. 2019; 105:669–680.
Article
37. Coorens M, Schneider VAF, de Groot AM, van Dijk A, Meijerink M, Wells JM, et al. Cathelicidins inhibit Escherichia coli-induced TLR2 and TLR4 activation in a viability-dependent manner. J Immunol. 2017; 199:1418–1428.
Article
38. Sørensen OE, Follin P, Johnsen AH, Calafat J, Tjabringa GS, Hiemstra PS, et al. Human cathelicidin, hCAP-18, is processed to the antimicrobial peptide LL-37 by extracellular cleavage with proteinase 3. Blood. 2001; 97:3951–3959.
Article
39. Lai Y, Adhikarakunnathu S, Bhardwaj K, Ranjith-Kumar CT, Wen Y, Jordan JL, et al. LL37 and cationic peptides enhance TLR3 signaling by viral double-stranded RNAs. PLoS One. 2011; 6:e26632.
Article
40. Lai Y, Yi G, Chen A, Bhardwaj K, Tragesser BJ, Rodrigo AV, et al. Viral double-strand RNA-binding proteins can enhance innate immune signaling by toll-like Receptor 3. PLoS One. 2011; 6:e25837.
Article
41. Wan M, van der Does AM, Tang X, Lindbom L, Agerberth B, Haeggström JZ. Antimicrobial peptide LL-37 promotes bacterial phagocytosis by human macrophages. J Leukoc Biol. 2014; 95:971–981.
Article
42. Wanke D, Mauch-Mucke K, Holler E, Hehlgans T. Human beta-defensin-2 and -3 enhance pro-inflammatory cytokine expression induced by TLR ligands via ATP-release in a P2X7R dependent manner. Immunobiology. 2016; 221:1259–1265.
Article
43. Semple F, MacPherson H, Webb S, Cox SL, Mallin LJ, Tyrrell C, et al. Human beta-defensin 3 affects the activity of pro-inflammatory pathways associated with MyD88 and TRIF. Eur J Immunol. 2011; 41:3291–3300.
Article
44. Khurshid Z, Naseem M, Yahya IAF, Mali M, Sannam Khan R, Sahibzada HA, et al. Significance and diagnostic role of antimicrobial cathelicidins (LL-37) peptides in oral health. Biomolecules. 2017; 7:pii: E80.
Article
45. Patel S, Akhtar N. Antimicrobial peptides (AMPs): The quintessential ‘offense and defense’ molecules are more than antimicrobials. Biomed Pharmacother. 2017; 95:1276–1283.
Article
46. Liu PT, Stenger S, Li H, Wenzel L, Tan BH, Krutzik SR, et al. Toll-like receptor triggering of a vitamin D-mediated human antimicrobial response. Science. 2006; 311:1770–1773.
Article
47. Liu PT, Schenk M, Walker VP, Dempsey PW, Kanchanapoomi M, Wheelwright M, et al. Convergence of IL-1beta and VDR activation pathways in human TLR2/1-induced antimicrobial responses. PLoS One. 2009; 4:e5810.
48. Boro M, Singh V, Balaji KN. Mycobacterium tuberculosis-triggered Hippo pathway orchestrates CXCL1/2 expression to modulate host immune responses. Sci Rep. 2016; 6:37695.
Article
49. Banoth B, Cassel SL. Mitochondria in innate immune signaling. Transl Res. 2018; 202:52–68.
Article
50. West AP, Brodsky IE, Rahner C, Woo DK, Erdjument-Bromage H, Tempst P, et al. TLR signalling augments macrophage bactericidal activity through mitochondrial ROS. Nature. 2011; 472:476–480.
Article
51. Geng J, Sun X, Wang P, Zhang S, Wang X, Wu H, et al. Kinases Mst1 and Mst2 positively regulate phagocytic induction of reactive oxygen species and bactericidal activity. Nat Immunol. 2015; 16:1142–1152.
Article
52. Min Y, Wi SM, Shin D, Chun E, Lee KY. Peroxiredoxin-6 negatively regulates bactericidal activity and NF-kappaB activity by interrupting TRAF6-ECSIT complex. Front Cell Infect Microbiol. 2017; 7:94.
Article
53. Oberkampf M, Guillerey C, Mouriès J, Rosenbaum P, Fayolle C, Bobard A, et al. Mitochondrial reactive oxygen species regulate the induction of CD8(+) T cells by plasmacytoid dendritic cells. Nat Commun. 2018; 9:2241.
Article
54. Shibutani ST, Saitoh T, Nowag H, Münz C, Yoshimori T. Autophagy and autophagy-related proteins in the immune system. Nat Immunol. 2015; 16:1014–1024.
Article
55. Kuballa P, Nolte WM, Castoreno AB, Xavier RJ. Autophagy and the immune system. Annu Rev Immunol. 2012; 30:611–646.
Article
56. Siqueira MDS, Ribeiro RM, Travassos LH. Autophagy and its interaction with intracellular bacterial pathogens. Front Immunol. 2018; 9:935.
Article
57. Gomes LC, Dikic I. Autophagy in antimicrobial immunity. Mol Cell. 2014; 54:224–233.
Article
58. Zhao YG, Zhang H. Autophagosome maturation: An epic journey from the ER to lysosomes. J Cell Biol. 2019; 218:757–770.
Article
59. Yin Z, Pascual C, Klionsky DJ. Autophagy: machinery and regulation. Microb Cell. 2016; 3:588–596.
Article
60. Katsuragi Y, Ichimura Y, Komatsu M. p62/SQSTM1 functions as a signaling hub and an autophagy adaptor. FEBS J. 2015; 282:4672–4678.
Article
61. Ying H, Yue BY. Optineurin: The autophagy connection. Exp Eye Res. 2016; 144:73–80.
Article
62. Viret C, Rozières A, Faure M. Novel insights into NDP52 autophagy receptor functioning. Trends Cell Biol. 2018; 28:255–257.
Article
63. Castrejón-Jimenèz NS, Leyva-Paredes K, Hernández-González JC, Luna-Herrera J, García-Pèrez BE. The role of autophagy in bacterial infections. Biosci Trends. 2015; 9:149–159.
Article
64. Watson RO, Manzanillo PS, Cox JS. Extracellular M. tuberculosis DNA targets bacteria for autophagy by activating the host DNA-sensing pathway. Cell. 2012; 150:803–815.
Article
65. Manzanillo PS, Ayres JS, Watson RO, Collins AC, Souza G, Rae CS, et al. The ubiquitin ligase parkin mediates resistance to intracellular pathogens. Nature. 2013; 501:512–516.
Article
66. Watson RO, Bell SL, MacDuff DA, Kimmey JM, Diner EJ, Olivas J, et al. The cytosolic sensor cGAS detects Mycobacterium tuberculosis DNA to induce type I interferons and activate autophagy. Cell Host Microbe. 2015; 17:811–819.
Article
67. Franco LH, Nair VR, Scharn CR, Xavier RJ, Torrealba JR, Shiloh MU, et al. The ubiquitin ligase Smurf1 functions in selective autophagy of Mycobacterium tuberculosis and anti-tuberculous host defense. Cell Host Microbe. 2017; 21:59–72.
Article
68. Chauhan S, Kumar S, Jain A, Ponpuak M, Mudd MH, Kimura T, et al. TRIMs and Galectins globally cooperate and TRIM16 and Galectin-3 co-direct autophagy in endomembrane damage homeostasis. Dev Cell. 2016; 39:13–27.
Article
69. Kumar S, Chauhan S, Jain A, Ponpuak M, Choi SW, Mudd M, et al. Galectins and TRIMs directly interact and orchestrate autophagic response to endomembrane damage. Autophagy. 2017; 13:1086–1087.
Article
70. Ponpuak M, Davis AS, Roberts EA, Delgado MA, Dinkins C, Zhao Z, et al. Delivery of cytosolic components by autophagic adaptor protein p62 endows autophagosomes with unique antimicrobial properties. Immunity. 2010; 32:329–341.
Article
71. Alonso S, Pethe K, Russell DG, Purdy GE. Lysosomal killing of Mycobacterium mediated by ubiquitin-derived peptides is enhanced by autophagy. Proc Natl Acad Sci U S A. 2007; 104:6031–6036.
Article
72. Yuk JM, Shin DM, Lee HM, Yang CS, Jin HS, Kim KK, et al. Vitamin D3 induces autophagy in human monocytes/macrophages via cathelicidin. Cell Host Microbe. 2009; 6:231–243.
Article
73. Rekha RS, Rao Muvva SS, Wan M, Raqib R, Bergman P, Brighenti S, et al. Phenylbutyrate induces LL-37-dependent autophagy and intracellular killing of Mycobacterium tuberculosis in human macrophages. Autophagy. 2015; 11:1688–1699.
Article
74. Høyer-Hansen M, Nordbrandt SP, Jäättelä M. Autophagy as a basis for the health-promoting effects of vitamin D. Trends Mol Med. 2010; 16:295–302.
Article
75. Chauhan S, Mandell MA, Deretic V. IRGM governs the core autophagy machinery to conduct antimicrobial defense. Mol Cell. 2015; 58:507–521.
Article
76. Kimmey JM, Huynh JP, Weiss LA, Park S, Kambal A, Debnath J, et al. Unique role for ATG5 in neutrophil-mediated immunopathology during M. tuberculosis infection. Nature. 2015; 528:565–569.
Article
77. Paik S, Kim JK, Chung C, Jo EK. Autophagy: a new strategy for host-directed therapy of tuberculosis. Virulence. 2018; 15:1–12.
Article
78. Stocks CJ, Schembri MA, Sweet MJ, Kapetanovic R. For when bacterial infections persist: Toll-like receptor-inducible direct antimicrobial pathways in macrophages. J Leukoc Biol. 2018; 103:35–51.
Article
79. Delgado MA, Elmaoued RA, Davis AS, Kyei G, Deretic V. Toll-like receptors control autophagy. EMBO J. 2008; 27:1110–1121.
Article
80. Delgado MA, Deretic V. Toll-like receptors in control of immunological autophagy. Cell Death Differ. 2009; 16:976–983.
Article
81. Xu Y, Jagannath C, Liu XD, Sharafkhaneh A, Kolodziejska KE, Eissa NT. Toll-like receptor 4 is a sensor for autophagy associated with innate immunity. Immunity. 2007; 27:135–144.
Article
82. Shin DM, Yuk JM, Lee HM, Lee SH, Son JW, Harding CV, et al. Mycobacterial lipoprotein activates autophagy via TLR2/1/CD14 and a functional vitamin D receptor signalling. Cell Microbiol. 2010; 12:1648–1665.
Article
83. Xu Y, Fattah EA, Liu XD, Jagannath C, Eissa NT. Harnessing of TLR-mediated autophagy to combat mycobacteria in macrophages. Tuberculosis (Edinb). 2013; 93:S33–S37.
Article
84. Chuang SY, Yang CH, Chou CC, Chiang YP, Chuang TH, Hsu LC. TLR-induced PAI-2 expression suppresses IL-1beta processing via increasing autophagy and NLRP3 degradation. Proc Natl Acad Sci U S A. 2013; 110:16079–16084.
Article
85. Lee JW, Nam H, Kim LE, Jeon Y, Min H, Ha S, et al. TLR4 (toll-like receptor 4) activation suppresses autophagy through inhibition of FOXO3 and impairs phagocytic capacity of microglia. Autophagy. 2018; DOI: 10.1080/15548627.2018.1556946. [Epub ahead of print].
Article
86. Franco LH, Fleuri AKA, Pellison NC, Quirino GFS, Horta CV, de Carvalho RVH, et al. Autophagy downstream of endosomal Toll-like receptor signaling in macrophages is a key mechanism for resistance to Leishmania major infection. J Biol Chem. 2017; 292:13087–13096.
Article
87. Campbell GR, Spector SA. Toll-like receptor 8 ligands activate a vitamin D mediated autophagic response that inhibits human immunodeficiency virus type 1. PLoS Pathog. 2012; 8:e1003017.
Article
88. Fujita K, Maeda D, Xiao Q, Srinivasula SM. Nrf2-mediated induction of p62 controls Toll-like receptor-4-driven aggresome-like induced structure formation and autophagic degradation. Proc Natl Acad Sci U S A. 2011; 108:1427–1432.
Article
89. Blanchet FP, Piguet V. Immunoamphisomes in dendritic cells amplify TLR signaling and enhance exogenous antigen presentation on MHC-II. Autophagy. 2010; 6:816–818.
Article
90. Li YY, Ishihara S, Aziz MM, Oka A, Kusunoki R, Tada Y, et al. Autophagy is required for toll-like receptor-mediated interleukin-8 production in intestinal epithelial cells. Int J Mol Med. 2011; 27:337–344.
Article
91. Peral de, Jones SA, Ni Cheallaigh C, Hearnden CA, Williams L, Winter J, et al. Autophagy regulates IL-23 secretion and innate T cell responses through effects on IL-1 secretion. J Immunol. 2012; 189:4144–4153.
Article
92. Shi CS, Kehrl JH. MyD88 and Trif target Beclin 1 to trigger autophagy in macrophages. J Biol Chem. 2008; 283:33175–33182.
Article
93. Shi CS, Kehrl JH. TRAF6 and A20 regulate lysine 63-linked ubiquitination of Beclin-1 to control TLR4-induced autophagy. Sci Signal. 2010; 3:ra42.
Article
94. Shi CS, Kehrl JH. Traf6 and A20 differentially regulate TLR4-induced autophagy by affecting the ubiquitination of Beclin 1. Autophagy. 2010; 6:986–987.
Article
95. Jabir MS, Ritchie ND, Li D, Bayes HK, Tourlomousis P, Puleston D, et al. Caspase-1 cleavage of the TLR adaptor TRIF inhibits autophagy and beta-interferon production during Pseudomonas aeruginosa infection. Cell Host Microbe. 2014; 15:214–227.
Article
96. Ní Cheallaigh C, Sheedy FJ, Harris J, Muñnoz-Wolf N, Lee J, West K, et al. A Common Variant in the Adaptor Mal Regulates Interferon Gamma Signaling. Immunity. 2016; 44:368–379.
Article
97. Yang Q, Liu TT, Lin H, Zhang M, Wei J, Luo WW, et al. TRIM32-TAX1BP1-dependent selective autophagic degradation of TRIF negatively regulates TLR3/4-mediated innate immune responses. PLoS Pathog. 2017; 13:e1006600.
Article
98. van der Vaart M, Korbee CJ, Lamers GE, Tengeler AC, Hosseini R, Haks MC, et al. The DNA damage-regulated autophagy modulator DRAM1 links mycobacterial recognition via TLR-MYD88 to autophagic defense. Cell Host Microbe. 2014; 15:753–767.
Article
99. Yang CS, Rodgers M, Min CK, Lee JS, Kingeter L, Lee JY, et al. The autophagy regulator Rubicon is a feedback inhibitor of CARD9-mediated host innate immunity. Cell Host Microbe. 2012; 11:277–289.
Article
100. Yang CS, Lee JS, Rodgers M, Min CK, Lee JY, Kim HJ, et al. Autophagy protein Rubicon mediates phagocytic NADPH oxidase activation in response to microbial infection or TLR stimulation. Cell Host Microbe. 2012; 11:264–276.
Article
101. Kim JH, Kim TH, Lee HC, Nikapitiya C, Uddin MB, Park ME, et al. Rubicon modulates antiviral type I interferon (IFN) signaling by targeting IFN regulatory factor 3 dimerization. J Virol. 2017; 91:e00248-17.
Article
Full Text Links
  • JBV
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr